Organoid > Volume 4; 2024 > Article
Lee and Lee: Advances in cardiac organoid technology: current trends and standardized quality assessment for toxicity testing in preclinical models

Abstract

Cardiotoxicity is a prominent cause of drug withdrawal, impacting drug development during preclinical and clinical phases, as well as post-market approval. Traditional 2-dimensional cell culture models, despite being resource-rich, suffer from a suboptimal physiological environment. Animal models provide an in vivo setting; however, their use is declining due to interspecies variability and ethical concerns. Cardiac organoids, which are derived from human induced pluripotent stem cells, incorporate human genetic information and contain various cell types within a 3-dimensional environment that mimics the human heart. As a result, they are becoming increasingly popular in the development of preclinical models for drug testing. To effectively use cardiac organoids as preclinical models, a standardized quality assessment is crucial. This review discusses current trends in cardiac organoid generation and examines recent analytical techniques for quality assessment.

Introduction

Toxicity is a primary cause of adverse drug reactions throughout preclinical and clinical drug development [1-3]. It also plays a significant role in the withdrawal of approved drugs, with cardiotoxicity accounting for 45% of these cases [4]. Drug withdrawals due to cardiotoxicity encompass a spectrum of phenomena, including cardiac arrhythmias, disturbances in electrophysical functions, myocardial ischemia, heart failures, myocardial necrosis, valvular heart disease, hypertension, and more [5-7]. Commonly used preclinical cell models have limitations, notably the absence of a physiological environment, relying predominantly on hERG (human ether-a-go-go-related gene) channel assays to detect cardiac arrhythmias [3,8]. Conversely, animal models offer the advantage of evaluating various types of cardiotoxicity in a physiological environment, but are subject to variations due to interspecies differences [9-11].
To overcome the limitations of these models, cardiac organoid models derived from human induced pluripotent stem cells (hiPSCs) have been proposed [12-17]. Cardiac organoids are composed of multiple cell types and are structured in a 3-dimensional (3D) environment that closely mimics the structure and environment of organs in humans [18]. The methods for generating hiPSC-derived cardiac organoids vary, leading to differences in the composition of cell types and their ratios based on the cell source and reagents used [19]. Therefore, to use the generated cardiac organoids as a cardiotoxicity model in drug development, it is essential to establish quality evaluation criteria [20]. This review presents the current trends in techniques for generating hiPSC-derived cardiac organoids and provides an overview of existing methods for quality analysis.

Trends in the development of cardiac organoids for toxicity testing

Ethics statement: This study was a literature review of previously published studies and was therefore exempt from institutional review board approval.
There are 2 main approaches for generating cardiac organoids using hiPSCs: the self-assembly method involving the formation of embryoid bodies and the reassembly of various 2-dimensional (2D) cell types at a specific ratio to generate a 3D structure [21-23]. Research trends in the differentiation of cardiac organoids from embryoid bodies are presented in Table 1 [14-17,24,25]. Hoang et al. [14] utilized a polydimethylsiloxane (PDMS)-based cell-patterning technique to induce self-organization of embryoid bodies of a specific size, leading to self-tissue organization and the development of a cardiac organoid model mimicking early heart development stages. Hofbauer et al. [15] showed that the regulation of the WNT/BMP signaling pathway facilitated lineage-specific self-tissue organization, resulting in cardiac organoids with structures resembling ventricles. These models exhibited ventricular-like structures and demonstrated heart regeneration and early features of heart disease when subjected to cryoinjury. Lewis-Israeli et al. [16] developed a cardiac organoid model with characteristics similar to human fetal heart employing a 3-step regulation of the WNT signaling pathway. This disease model was validated as reproducing congenital heart disease under conditions similar to maternal gestational diabetes. Drakhlis et al. [24] directly differentiated induced pluripotent stem cell aggregates embedded in Matrigel (Corning, New York, NY, USA), using a 2-step process for WNT signaling regulation, creating cardiac organoids resembling early developmental stages before heart tube formation. These organoid models replicated abnormal cardiac function phenotypes through NKX2.5 knockout. Lee et al. [17] developed a protocol for efficiently enhancing electrophysiological characteristics in cardiac organoids using ZM447439, an Aurora kinase inhibitor, during the differentiation process. Lee et al. [26] developed a protocol for chamber-formed cardiac organoids with cardiac structure and functionality using Matrigel to form embryoid bodies and a 2-step process for WNT signaling regulation. They confirmed vascularization through in vivo transplantation.
Methods for reassembling cardiac constituent cells into cardiac organoids within a 3D environment are presented in Table 2 [27-33]. Richards et al. [27] engineered organoids replicating the lumenized vascular network during developmental stages by incorporating adult stem cells to promote endothelial cell function, combined with existing myocardial cells, fibroblasts, and endothelial cells. Mills et al. [28] developed a 96-well plate with elastic pillars promoting functional enhancement by providing physical resistance during contraction, resulting in cardiac organoid formation around the pillars. They screened for culture conditions that increased maturity and elucidated the associated mechanisms. Li et al. [29] developed a human ventricle-like cardiac organoid chamber with a pumping chamber structure using a collagen-based hydrogel mold. Li et al. [30] developed a flexible mesh-shaped nanoelectrode array that, when seeded with a mixture of cardiac constituent cells, self-organized into 3D cardiac organoids. The bio-sensor that was distributed uniformly within the organoid enabled the analysis of electrophysiological properties in multiple organoids. In the study of Song et al. [31], cardiac midline-derived cells differentiated into spheroid-shape cardiac organoids through 2D differentiation, exhibiting more mature characteristics than organoids based on differentiated myocardial cells with involvement of the LEFTY-PITX2 signaling pathway. Meier et al. [25] developed self-organizing epicardioids with ventricular myocardium and epicardium using collagen I gel and retinoic acid signaling activation. The trends in cardiac organoid generation methods are gradually shifting towards techniques facilitating easier reconstitution of a consistent cell composition, particularly through lineage differentiation using embryoid bodies [21].

Trends in quality assessment methods for cardiac organoids

Quality assessments of cardiac organoids generated through various protocols are deemed imperative [23]. The methodologies proposed for evaluating the quality of cardiac organoids closely resemble those utilized for cardiomyocyte quality assessment. Established approaches include molecular analysis primarily based on genetics and structural characteristics, as well as functional analysis utilizing imaging techniques, as presented in Table 3 [34]. Table 3 summarizes the various methods used to assess the quality and characteristics of cardiac organoids, alongside their observed purposes and corresponding references [14-17,26-28,32,35-47].
Immunofluorescence techniques are employed to identify cell type markers, sarcomere arrangement, cardiac structure, interactions between cardiomyocytes and fibroblasts, and extracellular matrix composition. Polymerase chain reaction (PCR) and quantitative reverse transcription PCR (qRT-PCR) are used to evaluate cell type markers, function-related genes, mitochondrial biogenesis, and structure-related genes. Patch clamp and multi-electrode Array (MEA) assays are utilized to assess electrophysiological properties. Voltage-sensitive dye imaging and calcium imaging further contribute to understanding these properties. Optical coherence tomography (OCT) is applied for detailed structural and mitochondrial biogenesis analysis. Transmission electron microscopy (TEM) is used for structural analysis, including sarcomere length measurement. Hematoxylin-eosin staining assesses wall thickness, and image (video) analysis examines contractile properties. The seahorse mitochondrial stress test measures the oxygen consumption rate, providing insights into mitochondrial function. The terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay is used to detect apoptosis, while microscopy examines morphological characteristics. RNA in situ hybridization identifies cardiac lineage genes, and single-cell RNA sequencing (scRNA-seq) is used for comprehensive gene expression assays. Various advanced data analysis methods such as principal component analysis (PCA), gene set enrichment analysis (GSEA), differential gene expression analysis, gene set variation analysis, k-mean cluster analysis, and transcriptome analysis are conducted for in-depth data interpretation and validation of gene expression profiles. Proteomics is employed to study protein expression related to the adult heart.
This comprehensive overview highlights the range of techniques applied to assess the structural, functional, and molecular attributes of cardiac organoids, thereby providing a detailed framework for evaluating their quality and potential for research and clinical applications.
Recent advancements have introduced techniques that incorporate visual analysis and algorithmic assessments. Devarasetty et al. [48] developed software for visually analyzing video files of beating cardiac organoids and used it to analyze responsiveness to 5 different drugs affecting cardiac contractions. Sala et al. [49] developed an analysis tool based on imaging to measure the contraction and relaxation of muscular structures, nabling comparisons between single and multilayer cardiac cells, cardiac organoids, engineered cardiac tissues, and adult cardiomyocyte models. The tool’s applicability was confirmed in scenarios using various analysis tools, such as patch clamps. Feng et al. [35] produced chambered cardiac organoids and used RNA-seq analysis to identify the specific cellular composition of the fabricated organoids. Additionally, they developed an scRNA-seq data analysis method based on the random forest classification algorithm using machine learning. Kim et al. [50] developed low mechanical modulus and low impedance electrodes for electrophysiological measurements of 3D organoids, introducing a wet cardiac organoid analysis tool.
While these advanced techniques offer extensive information, they also come with notable limitations and challenges. Immunofluorescence is effective in identifying cell markers and structures, but is hindered by staining variability. PCR/qRT-PCR is useful for evaluating gene function, but requires high-quality RNA, which can be difficult to obtain. Patch clamp and MEA are valuable for measuring electrophysiological properties, but necessitate significant technical expertise and are susceptible to noise interference. TEM and OCT provide detailed structural analysis but may introduce artifacts during sample preparation. Seahorse assays are useful for assessing mitochondrial function, but are highly sensitive to environmental conditions. TUNEL assays are utilized to detect apoptosis but carry the risk of false positives. Lastly, advanced methods such as scRNA-seq, PCA, and GSEA yield comprehensive data, but are computationally intensive, costly, and require specialized expertise.
Additionally, as 3D structures, cardiac organoids pose challenges in traditional experimental approaches. These include difficulties in the adhesion of 3D organoids on 2D MEA plates, measurements of 3D structural characteristics using imaging techniques (e.g., TEM, video analysis), and reflection of the core of the organoid due to reagent permeability constraints (e.g., TUNEL assay, calcium imaging) [36,50-52]. To overcome these challenges, it is essential to develop analysis methods using computer algorithms or specialized analytical techniques tailored for 3D structures [53,54]. Furthermore, establishing quality assessment criteria that accurately reflect the structural characteristics of cardiac organoids is crucial for advancing their effective application.

Conclusion

In conclusion, the production of hiPSC-derived cardiac organoids exhibits variability depending on factors such as cell type, differentiation media conditions, and more [34,55]. Two primary methods, involving the formation of embryoid bodies and the reassembling of 2D heart constituent cells, are employed based on the initial differentiation approach [21,56,57]. The embryoid body formation method offers the advantage of being composed of cells derived from the same origin [21]. However, the reassembly method provides high reproducibility of relative control cell ratios but has a weaker physical structure for heart development [21]. Consequently, quality assessment is essential for the effective utilization of generated cardiac organoids in preclinical evaluations. Various experimental methods are used to assess the quality of produced cardiac organoids, and more suitable experimental methods for organoid quality are being developed. With more accurate and precise quality assessments, cardiac organoids can exhibit structural and physiological reactivity similar to the human heart. These organoids can be used not only as preclinical cardiotoxicity models, but also as models for cardiac development research and patient-specific treatments.

NOTES

Conflict of interest

No potential conflict of interest relevant to this article was reported.

Funding

This work was supported by the Technology Innovation Program (#20009774) funded by the Ministry of Trade, Industry, and Energy (MOTIE, Korea) and by grants from the National Research Foundation of Korea (NRF-2022M3A9H1015784, RS-2023-00225239).

Data availability

Please contact the corresponding author for any inquiries related to data availability.

Table 1.
Protocols for cardiac organoid differentiation from embryoid bodies
Research group (year) Differentiation platform Stem cells
Differentiation
Reference
Source Media WNT regulators Factors Media Cell number
Syracuse University (2021) Customized microfabricated wells hiPSCs mTeSR1 10 μM CHIR99021, 5 μM IWP4 RPMI 1640 medium+B27 supplement -insulin 0.63×105 cells/cm2 [14]
Vienna BioCenter (2021) Ultra-low attachment 96-well plates hESCs, hiPSCs E8 4-9 μM CHIR99021, 5 μM IWP2 8-30 ng/mL FGF2, 4-50 ng/mL activin A, 10 ng/mL BMP4, 1 or 10 μg/mL Insulin, 0.5 μM retinoic acid, 5 μM LY294002 CDM medium 2.5×104-7.5×104 cells/well [15]
Michigan State University (2021) Round-bottom ultra-low attachment 96-well plates hiPSCs E8+1% penicillin/streptomycin 4, 2 μM CHIR99021, 2 μM C59 1.25 ng/mL BMP4, 1ng/mL activin A RPMI 1640 medium+B27 supplement -insulin 1.0×104 cells/well [16]
Hannover Medical School (2021) U-shaped ultra-low attachment 96-well plate hESCs E8 7.5 μM CHIR99021, 5 μM IWP2 RPMI 1640 medium+B27 supplement -insulin 0.5×104 cells/well [24]
Korea Institute of Toxicology (2021) Ultra-low attachment 6-well plates hiPSCs mTeSR1 10 μM CHIR99021, 5 μM IWP2 0.2 mg/mL L-ascorbic acid, 100 nM ZM447439 RPMI 1640 medium+B27 supplement -insulin 1×106 cells/well [17]
Technical University of Munich (2023) U-shaped 96-well plates hiPSCs E8 1.5 μM CHIR-99021, 5 μM IWP2 10 ng/mL BPM4, 50 ng/mL activin A, 30 ng/mL bFGF, 5 μM LY-29004 DMEM/F-12 medium+IMDM 0.3×104-0.4×104 cells/well [25]

hiPSC, human induced pluripotent stem cell; hESC, human embryonic stem cell.

Table 2.
Protocols for cardiac organoid generation from reassembling cardiac constituent cells
Research group (year) Aggregation platform Cell composition Cell number Reference
Clemson University (2017) Agarose hydrogel mold 55% hiPSC-CMs, 24% hFBs, 14% HUVECs, and 7% hADSCs ~2.0×106 cells/mL [27]
The University of Queensland (2017) 96-well plate with wells containng 2 elastomeric posts made with PDMS hiPSC-CMs (~70% α-actinin+/CTNT+), CD90+ stromal cells 5×104 cardiac cells [28]
Novoheart Limited (2018) Pumping chamber structure scaffold with collagen-based hydrogel 90% ESC-CMs (cTNT+ /MLC2v+), 10% dermal FB 1×107 CMs+1×106 FBs [29]
ACECR (2019) Matrigel-coated 24-well plate 50% CPCs, 25% MSCs, 25% ECs [32]
Harvard University (2019) Matrigel-coated stretchable mesh nanoeletronics hiPSC-CPCs, hMSCs 2-3×106 hiPSC-CPCs+ 2-4×105 hMSCs [30]
The University of Texas at El Paso (2022) 3D printed annular ring-like scaffolds of hydrogel (gelatin-alginate) constructs CMs, CFs, ECs [33]
Korea University (2021) Poly (2-hydroxyethyl methacrylate)-coated 6-well plate hESC-CMCs 2-3×105 cells/cm2 [31]

hiPSC, human induced pluripotent stem cell; CM, cardiomyocyte; hFB, human fibroblast; HUVEC, human umbilical vein endothelial cell; hADSC, human adipose-derived stem cell; PDMS, polydimethylsiloxane; FB, fibroblast; CPC, cardiac progenitor cell; EC, endothelial cell; CF, cardiac fibroblast; MSC, mesenchymal stem cell; hMSC, human MSC; hESC, human embryonic stem cell; 3D, 3-dimensional; CMC, cardiac mesoderm cel.

Table 3.
Methods and analysis for quality assessment
Methods Observed purpose Reference
Immunofluorescence Cell type marker [14, 16, 17, 26-28, 37-40]
Sarcomere arrangement [39, 40]
Cardiac structure [15, 16, 35, 40]
Interaction between cardiomyocyte and fibroblast [40, 41]
Extracellular matrix composition [27]
PCR/quantitative reverse transcription PCR Cell type marker [17, 32, 38]
Function-related gene [15, 16, 39, 40, 42]
Mitochondria biogenesis [26]
Structure-related gene [15, 41]
Patch clamp Electrophysiological properties [14, 16, 26-28, 32, 35, 36, 38-44]
Microelectrode array
Voltage-sensitive dye imaging
Calcium imaging
OCT
Transmission electron microscopy Structural analysis [15, 16, 32, 37, 40, 43]
Sarcomere length [26, 28, 40, 42, 43]
Hematoxylin-eosin staining Wall thickness [32, 42]
Image (video) analysis Contractile properties [27, 28]
Seahorse mitochondrial stress test Oxygen consumption rate [16, 28, 40, 45]
TUNEL assay Apoptosis [27, 41, 42, 45]
Microscopy Morphology [26, 38, 42, 43, 45]
RNA in situ hybridization Cardiac lineage genes [43]
Single-cell RNA sequencing Gene expression assay [15, 26, 35, 38, 40, 43, 45-47]
Principal component analysis
Gene set enrichment analysis
Differential gene expression analysis
Gene set variation analysis
k-mean cluster analysis
Transcriptome analysis
Proteomics Protein expression related to adult heart [15, 28, 46]
OCT Mitochondria biogenesis [16]

PCR, polymerase chain reaction; OCT, optical coherence tomography; TUNEL, terminal deoxynucleotidyl transferase dUTP nick-end labeling.

References

1. Laverty H, Benson C, Cartwright E, Cross M, Garland C, Hammond T, et al. How can we improve our understanding of cardiovascular safety liabilities to develop safer medicines? Br J Pharmacol 2011;163:675-93.
crossref pmid pmc
2. Hunter FMI, Bento AP, Bosc N, Gaulton A, Hersey A, Leach AR. Drug safety data curation and modeling in ChEMBL: boxed warnings and withdrawn drugs. Chem Res Toxicol 2021;34:385-95.
crossref pmid pmc
3. Pognan F, Beilmann M, Boonen HC, Czich A, Dear G, Hewitt P, et al. The evolving role of investigative toxicology in the pharmaceutical industry. Nat Rev Drug Discov 2023;22:317-35.
crossref pmid pmc pdf
4. Stevens JL, Baker TK. The future of drug safety testing: expanding the view and narrowing the focus. Drug Discov Today 2009;14:162-7.
crossref pmid
5. Seltzer JH, Gintant G, Amiri-Kordestani L, Singer J, Koplowitz LP, Moslehi JJ, et al. Assessing cardiac safety in oncology drug development. Am Heart J 2019;214:125-33.
crossref pmid pmc
6. Mamoshina P, Rodriguez B, Bueno-Orovio A. Toward a broader view of mechanisms of drug cardiotoxicity. Cell Rep Med 2021;2:100216.
crossref pmid pmc
7. Kocadal K, Saygi S, Alkas FB, Sardas S. Drug-associated cardiovascular risks: a retrospective evaluation of withdrawn drugs. North Clin Istanb 2018;6:196-202.
crossref pmid pmc
8. Priest BT, Bell IM, Garcia ML. Role of hERG potassium channel assays in drug development. Channels (Austin) 2008;2:87-93.
crossref pmid
9. Lamberti M, Giovane G, Garzillo EM, Avino F, Feola A, Porto S, et al. Animal models in studies of cardiotoxicity side effects from antiblastic drugs in patients and occupational exposed workers. Biomed Res Int 2014;2014:240642.
crossref pmid pmc pdf
10. Meng C, Fan L, Wang X, Wang Y, Li Y, Pang S, et al. Preparation and evaluation of animal models of cardiotoxicity in antineoplastic therapy. Oxid Med Cell Longev 2022;2022:3820591.
crossref pmid pmc pdf
11. Daley MC, Mende U, Choi BR, McMullen PD, Coulombe KL. Beyond pharmaceuticals: Fit-for-purpose new approach methodologies for environmental cardiotoxicity testing. ALTEX 2023;40:103-16.
crossref pmid
12. Nugraha B, Buono MF, Emmert MY. Modelling human cardiac diseases with 3D organoid. Eur Heart J 2018;39:4234-7.
crossref pmid
13. Muniyandi P, O’Hern C, Popa MA, Aguirre A. Biotechnological advances and applications of human pluripotent stem cell-derived heart models. Front Bioeng Biotechnol 2023;11:1214431.
crossref pmid pmc
14. Hoang P, Kowalczewski A, Sun S, Winston TS, Archilla AM, Lemus SM, et al. Engineering spatial-organized cardiac organoids for developmental toxicity testing. Stem Cell Reports 2021;16:1228-44.
crossref pmid pmc
15. Hofbauer P, Jahnel SM, Papai N, Giesshammer M, Deyett A, Schmidt C, et al. Cardioids reveal self-organizing principles of human cardiogenesis. Cell 2021;184:3299-317.
crossref pmid
16. Lewis-Israeli YR, Wasserman AH, Gabalski MA, Volmert BD, Ming Y, Ball KA, et al. Self-assembling human heart organoids for the modeling of cardiac development and congenital heart disease. Nat Commun 2021;12:5142.
crossref pmid pmc pdf
17. Lee SJ, Kim HA, Kim SJ, Lee HA. Improving generation of cardiac organoids from human pluripotent stem cells using the aurora kinase inhibitor ZM447439. Biomedicines 2021;9:1952.
crossref pmid pmc
18. Zhao Z, Chen X, Dowbaj AM, Sljukic A, Bratlie K, Lin L, et al. Organoids. Nat Rev Methods Primers 2022;2:94.
crossref pmid pmc
19. Roshanravan N, Ghaffari S, Bastani S, Pahlavan S, Asghari S, Doustvandi MA, et al. Human cardiac organoids: a recent revolution in disease modeling and regenerative medicine. J Cardiovasc Thorac Res 2023;15:68-72.
crossref pmid pmc pdf
20. Kim J, Koo BK, Knoblich JA. Human organoids: model systems for human biology and medicine. Nat Rev Mol Cell Biol 2020;21:571-84.
crossref pmid pmc pdf
21. Lewis-Israeli YR, Wasserman AH, Aguirre A. Heart organoids and engineered heart tissues: novel tools for modeling human cardiac biology and disease. Biomolecules 2021;11:1277.
crossref pmid pmc
22. Sahara M. Recent advances in generation of in vitro cardiac organoids. Int J Mol Sci 2023;24:6244.
crossref pmid pmc
23. Wu F, He Q, Li F, Yang X. A review of protocols for engineering human cardiac organoids. Heliyon 2023;9:e19938.
crossref pmid pmc
24. Drakhlis L, Biswanath S, Farr CM, Lupanow V, Teske J, Ritzenhoff K, et al. Human heart-forming organoids recapitulate early heart and foregut development. Nat Biotechnol 2021;39:737-46.
crossref pmid pmc pdf
25. Meier AB, Zawada D, De Angelis MT, Martens LD, Santamaria G, Zengerle S, et al. Epicardioid single-cell genomics uncovers principles of human epicardium biology in heart development and disease. Nat Biotechnol 2023;41:1787-800.
crossref pmid pmc pdf
26. Lee SG, Kim YJ, Son MY, Oh MS, Kim J, Ryu B, et al. Generation of human iPSCs derived heart organoids structurally and functionally similar to heart. Biomaterials 2022;290:121860.
crossref pmid
27. Richards DJ, Coyle RC, Tan Y, Jia J, Wong K, Toomer K, et al. Inspiration from heart development: Biomimetic development of functional human cardiac organoids. Biomaterials 2017;142:112-23.
crossref pmid pmc
28. Mills RJ, Titmarsh DM, Koenig X, Parker BL, Ryall JG, Quaife-Ryan GA, et al. Functional screening in human cardiac organoids reveals a metabolic mechanism for cardiomyocyte cell cycle arrest. Proc Natl Acad Sci U S A 2017;114:E8372-81.
crossref pmid pmc
29. Li RA, Keung W, Cashman TJ, Backeris PC, Johnson BV, Bardot ES, et al. Bioengineering an electro-mechanically functional miniature ventricular heart chamber from human pluripotent stem cells. Biomaterials 2018;163:116-27.
crossref pmid pmc
30. Li Q, Nan K, Le Floch P, Lin Z, Sheng H, Blum TS, et al. Cyborg Organoids: implantation of nanoelectronics via organogenesis for tissue-wide electrophysiology. Nano Lett 2019;19:5781-9.
crossref pmid
31. Song MH, Choi SC, Noh JM, Joo HJ, Park CY, Cha JJ, et al. LEFTY-PITX2 signaling pathway is critical for generation of mature and ventricular cardiac organoids in human pluripotent stem cell-derived cardiac mesoderm cells. Biomaterials 2021;278:121133.
crossref pmid
32. Varzideh F, Pahlavan S, Ansari H, Halvaei M, Kostin S, Feiz MS, et al. Human cardiomyocytes undergo enhanced maturation in embryonic stem cell-derived organoid transplants. Biomaterials 2019;192:537-50.
crossref pmid
33. Alonzo M, El Khoury R, Nagiah N, Thakur V, Chattopadhyay M, Joddar B. 3D biofabrication of a cardiac tissue construct for sustained longevity and function. ACS Appl Mater Interfaces 2022;14:21800-13.
crossref pmid pmc pdf
34. Kim H, Kamm RD, Vunjak-Novakovic G, Wu JC. Progress in multicellular human cardiac organoids for clinical applications. Cell Stem Cell 2022;29:503-14.
crossref pmid pmc
35. Feng W, Schriever H, Jiang S, Bais A, Wu H, Kostka D, Li G. Computational profiling of hiPSC-derived heart organoids reveals chamber defects associated with NKX2-5 deficiency. Commun Biol 2022;5:399.
crossref pmid pmc pdf
36. Lyu Q, Gong S, Lees JG, Yin J, Yap LW, Kong AM, et al. A soft and ultrasensitive force sensing diaphragm for probing cardiac organoids instantaneously and wirelessly. Nat Commun 2022;13:7259.
crossref pmid pmc pdf
37. Voges HK, Mills RJ, Elliott DA, Parton RG, Porrello ER, Hudson JE. Development of a human cardiac organoid injury model reveals innate regenerative potential. Development 2017;144:1118-27.
crossref pmid pdf
38. Ho BX, Pang JK, Chen Y, Loh YH, An O, Yang HH, et al. Robust generation of human-chambered cardiac organoids from pluripotent stem cells for improved modelling of cardiovascular diseases. Stem Cell Res Ther 2022;13:529.
crossref pmid pmc pdf
39. Ghosheh M, Ehrlich A, Ioannidis K, Ayyash M, Goldfracht I, Cohen M, et al. Electro-metabolic coupling in multi-chambered vascularized human cardiac organoids. Nat Biomed Eng 2023;7:1493-513.
crossref pmid pdf
40. Giacomelli E, Meraviglia V, Campostrini G, Cochrane A, Cao X, van Helden RW, et al. Human-iPSC-derived cardiac stromal cells enhance maturation in 3D cardiac microtissues and reveal non-cardiomyocyte contributions to heart disease. Cell Stem Cell 2020;26:862-79.
crossref pmid pmc
41. Patino-Guerrero A, Ponce Wong RD, Kodibagkar VD, Zhu W, Migrino RQ, Graudejus O, et al. Development and characterization of isogenic cardiac organoids from human-induced pluripotent stem cells under supplement starvation regimen. ACS Biomater Sci Eng 2023;9:944-58.
crossref pmid pdf
42. Kahn-Krell A, Pretorius D, Guragain B, Lou X, Wei Y, Zhang J, et al. A three-dimensional culture system for generating cardiac spheroids composed of cardiomyocytes, endothelial cells, smooth-muscle cells, and cardiac fibroblasts derived from human induced-pluripotent stem cells. Front Bioeng Biotechnol 2022;10:908848.
crossref pmid pmc
43. Silva AC, Matthys OB, Joy DA, Kauss MA, Natarajan V, Lai MH, et al. Co-emergence of cardiac and gut tissues promotes cardiomyocyte maturation within human iPSC-derived organoids. Cell Stem Cell 2021;28:2137-52.
crossref pmid
44. Ming Y, Hao S, Wang F, Lewis-Israeli YR, Volmert BD, Xu Z, et al. Longitudinal morphological and functional characterization of human heart organoids using optical coherence tomography. Biosens Bioelectron 2022;207:114136.
crossref pmid pmc
45. Richards DJ, Li Y, Kerr CM, Yao J, Beeson GC, Coyle RC, et al. Human cardiac organoids for the modelling of myocardial infarction and drug cardiotoxicity. Nat Biomed Eng 2020;4:446-62.
crossref pmid pmc pdf
46. Mills RJ, Parker BL, Quaife-Ryan GA, Voges HK, Needham EJ, Bornot A, et al. Drug screening in human PSC-cardiac organoids identifies pro-proliferative compounds acting via the mevalonate pathway. Cell Stem Cell 2019;24:895-907.
crossref pmid
47. Voges HK, Foster SR, Reynolds L, Parker BL, Devilée L, Quaife-Ryan GA, et al. Vascular cells improve functionality of human cardiac organoids. Cell Rep 2023;42:112322.
crossref pmid
48. Devarasetty M, Forsythe S, Shupe TD, Soker S, Bishop CE, Atala A, et al. Optical tracking and digital quantification of beating behavior in bioengineered human cardiac organoids. Biosensors (Basel) 2017;7:24.
crossref pmid pmc
49. Sala L, van Meer BJ, Tertoolen LG, Bakkers J, Bellin M, Davis RP, et al. MUSCLEMOTION: a versatile open software tool to quantify cardiomyocyte and cardiac muscle contraction in vitro and in vivo. Circ Res 2018;122:e5-16.
crossref pmid pmc
50. Kim M, Hwang JC, Min S, Park YG, Kim S, Kim E, et al. Multimodal characterization of cardiac organoids using integrations of pressure-sensitive transistor arrays with three-dimensional liquid metal electrodes. Nano Lett 2022;22:7892-901.
crossref pmid pmc pdf
51. Park Y, Franz CK, Ryu H, Luan H, Cotton KY, Kim JU, et al. Three-dimensional, multifunctional neural interfaces for cortical spheroids and engineered assembloids. Sci Adv 2021;7:eabf9153.
crossref pmid pmc
52. Chung WG, Kim E, Song H, Lee J, Lee S, Lim K, et al. Recent advances in electrophysiological recording platforms for brain and heart organoids. Adv NanoBiomed Res 2022;2:2200081.
crossref pdf
53. Cai C, Guo P, Zhou Y, Zhou J, Wang Q, Zhang F, et al. Deep learning-based prediction of drug-induced cardiotoxicity. J Chem Inf Model 2019;59:1073-84.
crossref pmid pmc
54. Matsui T, Shinozawa T. Human organoids for predictive toxicology research and drug development. Front Genet 2021;12:767621.
crossref pmid pmc
55. Ramirez-Calderon G, Colombo G, Hernandez-Bautista CA, Astro V, Adamo A. Heart in a dish: from traditional 2D differentiation protocols to cardiac organoids. Front Cell Dev Biol 2022;10:855966.
crossref pmid pmc
56. Martin M, Gähwiler EK, Generali M, Hoerstrup SP, Emmert MY. Advances in 3D organoid models for stem cell-based cardiac regeneration. Int J Mol Sci 2023;24:5188.
crossref pmid pmc
57. Brassard JA, Lutolf MP. Engineering stem cell self-organization to build better organoids. Cell Stem Cell 2019;24:860-76.
crossref pmid
TOOLS
METRICS Graph View
  • 0 Crossref
  •  0 Scopus
  • 2,189 View
  • 27 Download
ORCID iDs

Su-Jin Lee
https://orcid.org/0000-0003-1266-2522

Hyang-Ae Lee
https://orcid.org/0000-0002-0036-4555

Related articles


ABOUT
BROWSE ARTICLES
EDITORIAL POLICY
FOR CONTRIBUTORS
Editorial Office
Room 319, Hall 1 of Chonbuk National University Dental College, 20, Geonji-ro, Deokjin-gu, Jeonju 54907, Korea
Tel: +82-63-270-4024    E-mail: editor@j-organoid.org                

Copyright © 2024 by The Organoid Society.

Developed in M2PI

Close layer
prev next